Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Neurosurg Focus ; 32(4): E8, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22463118

RESUMO

Intracerebral hemorrhage (ICH) is a subtype of stoke that may cause significant morbidity and mortality. Brain injury due to ICH initially occurs within the first few hours as a result of mass effect due to hematoma formation. However, there is increasing interest in the mechanisms of secondary brain injury as many patients continue to deteriorate clinically despite no signs of rehemorrhage or hematoma expansion. This continued insult after primary hemorrhage is believed to be mediated by the cytotoxic, excitotoxic, oxidative, and inflammatory effects of intraparenchymal blood. The main factors responsible for this injury are thrombin and erythrocyte contents such as hemoglobin. Therapies including thrombin inhibitors, N-methyl-D-aspartate antagonists, chelators to bind free iron, and antiinflammatory drugs are currently under investigation for reducing this secondary brain injury. This review will discuss the molecular mechanisms of brain injury as a result of intraparenchymal blood, potential targets for therapeutic intervention, and treatment strategies currently in development.


Assuntos
Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patologia , Hemina/fisiologia , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Trombina/fisiologia , Antitrombinas/uso terapêutico , Hemorragia Cerebral/complicações , Hemina/antagonistas & inibidores , Hemina/metabolismo , Humanos , Quelantes de Ferro/uso terapêutico , Vias Neurais/metabolismo , Vias Neurais/patologia , Vias Neurais/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Acidente Vascular Cerebral/etiologia , Trombina/antagonistas & inibidores , Trombina/metabolismo
2.
Mol Ther ; 16(8): 1366-71, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18560415

RESUMO

Glycogen storage disease type II (Pompe disease; MIM 232300) stems from the deficiency of acid alpha-glucosidase (GAA; acid maltase; EC 3.2.1.20), which primarily involves cardiac and skeletal muscles. An adeno-associated virus 2/8 (AAV2/8) vector containing the muscle creatine kinase (MCK) (CK1) reduced glycogen content by approximately 50% in the heart and quadriceps in GAA-knockout (GAA-KO) mice; furthermore, an AAV2/8 vector containing the hybrid alpha-myosin heavy chain enhancer-/MCK enhancer-promoter (MHCK7) cassette reduced glycogen content by >95% in heart and >75% in the diaphragm and quadriceps. Transduction with an AAV2/8 vector was higher in the quadriceps than in the gastrocnemius. An AAV2/9 vector containing the MHCK7 cassette corrected GAA deficiency in the distal hindlimb, and glycogen accumulations were substantially cleared by human GAA (hGAA) expression therein; however, the analogous AAV2/7 vector achieved much lower efficacy. Administration of the MHCK7-containing vectors significantly increased striated muscle function as assessed by increased Rotarod times at 18 weeks after injection, whereas the CK1-containing vector did not increase Rotarod performance. Importantly, type IIb myofibers in the extensor digitalis longus (EDL) were transduced, thereby correcting a myofiber type that is unresponsive to enzyme replacement therapy. In summary, AAV8 and AAV9-pseudotyped vectors containing the MHCK7 regulatory cassette achieved enhanced efficacy in Pompe disease mice.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Doença de Depósito de Glicogênio Tipo II/terapia , Músculo Estriado/metabolismo , Animais , Creatina Quinase Forma MM/genética , Creatina Quinase Forma MM/metabolismo , Elementos Facilitadores Genéticos/genética , Feminino , Vetores Genéticos/genética , Glicogênio/metabolismo , Doença de Depósito de Glicogênio Tipo II/genética , Membro Posterior/metabolismo , Membro Posterior/patologia , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Estriado/enzimologia , Músculo Estriado/patologia , Miocárdio/metabolismo , Cadeias Pesadas de Miosina/genética , Regiões Promotoras Genéticas/genética , Músculo Quadríceps/metabolismo , Músculo Quadríceps/patologia , Transdução Genética , alfa-Glucosidases/genética , alfa-Glucosidases/metabolismo
3.
Int J Oncol ; 52(1): 47-54, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29115565

RESUMO

Glioblastoma multiforme (GBM) is one of the most common primary malignant brain tumors. Unraveling the molecular and genetic complexity that determines GBM's pronounced migratory property could provide new options for therapeutic targeting that may significantly complement current surgical and chemoradiation therapy and alter the current poor outcome. In this study, we establish stable AJAP1 overexpressing glioma cells in order to examine in vivo tumor growth. We examine AJAP1 localization by confocal microscopy and AJAP1's functional effect on migration and invasion across surfaces coated with laminin. Finally, analysis of AJAP1 expression in murine xenografts and GBM primary tumors revealed its association with tumor growth and survival. Stable overexpression of AJAP1 promotes adherence, decreases invasion of glioma cells through an extracellular-like matrix, and slows migration in the presence of laminin. These observations are reversed by gene knockdown using multiple siRNAs. Additionally, overexpression of AJAP1 decreases colony formation in glioma cells, and leads to smaller tumor growth with increased survival in glioma xenograft mice. Loss of AJAP1 protein expression predicts worse survival in GBM patients. AJAP1 overexpression decreases cell motility in the presence of laminin and decreases tumor growth in xenografts. Its loss of expression predicts worse survival in patients. This study extends our prior observations and implicates AJAP1 as a potential prognostic marker and a viable target for therapeutic intervention in GBM.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Moléculas de Adesão Celular/biossíntese , Movimento Celular/fisiologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Animais , Apoptose/fisiologia , Neoplasias Encefálicas/genética , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Processos de Crescimento Celular/fisiologia , Membrana Celular/metabolismo , Técnicas de Silenciamento de Genes , Glioblastoma/genética , Xenoenxertos , Humanos , Camundongos , Invasividade Neoplásica , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética
4.
Cancer Res ; 65(3): 919-24, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15705891

RESUMO

Through digital karyotyping of permanent medulloblastoma cell lines, we found that the homeobox gene OTX2 was amplified more than 10-fold in three cell lines. Gene expression analyses showed that OTX2 transcripts were present at high levels in 14 of 15 (93%) medulloblastomas with anaplastic histopathologic features. Knockdown of OTX2 expression by siRNAs inhibited medulloblastoma cell growth in vitro, whereas pharmacologic doses of all-trans retinoic acid repressed OTX2 expression and induced apoptosis only in medulloblastoma cell lines that expressed OTX2. These observations suggest that OTX2 is essential for the pathogenesis of anaplastic medulloblastomas and that these tumors may be amenable to therapy with all-trans-retinoic acid.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/genética , Proteínas de Homeodomínio/genética , Meduloblastoma/tratamento farmacológico , Meduloblastoma/genética , Proteínas do Tecido Nervoso/genética , Transativadores/genética , Tretinoína/farmacologia , Neoplasias Encefálicas/metabolismo , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Amplificação de Genes , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/biossíntese , Humanos , Meduloblastoma/metabolismo , Meduloblastoma/patologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/biossíntese , Oncogenes/efeitos dos fármacos , Oncogenes/genética , Fatores de Transcrição Otx , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Transativadores/antagonistas & inibidores , Transativadores/biossíntese
5.
Life Sci ; 78(6): 614-21, 2006 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-16137713

RESUMO

Chemokine-like factor 1 (CKLF1) exhibits chemotactic effects on leukocytes. Its amino acid sequence shares similarity with those of TARC/CCL17 and MDC/CCL22, the cognate ligands for CCR4. The chemotactic effects of CKLF1 for CCR4-transfected cells could be desensitized by TARC/CCL17 and markedly inhibited by PTX. CKLF1 induced a calcium flux in CCR4-transfected cells and fully desensitized a subsequent response to TARC/CCL17, and TARC/CCL17 could partly desensitize the response to CKLF1. CKLF1 caused significant receptor internalization in pCCR4-EGFP transfected cells. Taken together, CKLF1 is a novel functional ligand for CCR4.


Assuntos
Quimiocinas/farmacologia , Receptores de Quimiocinas/metabolismo , Cálcio/metabolismo , Células Cultivadas , Quimiocina CCL17 , Quimiocinas CC/farmacologia , Quimiotaxia , Humanos , Ligantes , Proteínas com Domínio MARVEL , Receptores CCR4 , Proteínas Recombinantes/farmacologia , Transfecção
6.
Cancer Res ; 64(15): 5048-50, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15289301

RESUMO

The phosphatidylinositol 3'-kinase pathway is activated in multiple advanced cancers, including glioblastomas, through inactivation of the PTEN tumor suppressor gene. Recently, mutations in PIK3CA, a member of the family of phosphatidylinositol 3'-kinase catalytic subunits, were identified in a significant fraction (25-30%) of colorectal cancers, gastric cancers, and glioblastomas and in a smaller fraction of breast and lung cancers. These mutations were found to cluster into two major "hot spots" located in the helical and catalytic domains. To determine whether PIK3CA is genetically altered in brain tumors, we performed a large-scale mutational analysis of the helical and catalytic domains. A total of 13 mutations of PIK3CA within these specific domains were identified in anaplastic oligodendrogliomas, anaplastic astrocytomas, glioblastoma multiforme, and medulloblastomas, whereas no mutations were identified in ependymomas or low-grade astrocytomas. These observations implicate PIK3CA as an oncogene in a wider spectrum of adult and pediatric brain tumors and suggest that PIK3CA may be a useful diagnostic marker or a therapeutic target in these cancers.


Assuntos
Astrocitoma/genética , Neoplasias Encefálicas/genética , Meduloblastoma/genética , Mutação , Oligodendroglioma/genética , Fosfatidilinositol 3-Quinases/genética , Astrocitoma/patologia , Neoplasias Encefálicas/patologia , Classe I de Fosfatidilinositol 3-Quinases , DNA de Neoplasias/genética , Humanos , Meduloblastoma/patologia , Oligodendroglioma/patologia , Monoéster Fosfórico Hidrolases/genética
7.
Biochim Biophys Acta ; 1591(1-3): 163-173, 2002 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-12183067

RESUMO

Chemokine-like factor 1 (CKLF1) is a novel cytokine first cloned from U937 cells. It contains different splicing forms and has chemotactic effects on a wide spectrum of cells both in vitro and in vivo; it can also stimulate the regeneration of skeletal muscle cells in vivo, but the mechanism remains unclear. To probe the myogenesis function of CKLF2, which is the largest isoform of CKLFs, C2C12 murine myoblasts were stably transfected with human CKLF2 eukaryotic expression vector. Compared with control vector transfected C2C12 cells, CKLF2 overexpression causes accelerated myoblast proliferation as determined by cell counting and [(3)H]TdR incorporation assays. In addition, CKLF2 overexpression also promotes cell differentiation, which was determined by higher expression levels of myogenin, creatine kinase, myosin and the accelerated myoblast fusion. Further analysis also indicates that CKLF2 could activate the transcription activity of the bHLH/MyoD and MEF2 families. Finally, DNA synthesis and myotube formation could also be promoted by growing C2C12 cells in conditioned media from CKLF2-transfected cells. These findings strongly suggest a role for human CKLF2 in regulation of skeletal muscle myogenesis.


Assuntos
Diferenciação Celular/fisiologia , Quimiocinas/fisiologia , Músculo Esquelético/citologia , Isoformas de Proteínas/fisiologia , Animais , Miosinas Cardíacas/metabolismo , Divisão Celular/fisiologia , Linhagem Celular , Sobrevivência Celular/fisiologia , Quimiocinas/biossíntese , Creatina Quinase/metabolismo , Creatina Quinase Forma MM , Proteínas de Ligação a DNA/genética , Isoenzimas/metabolismo , Proteínas com Domínio MARVEL , Fatores de Transcrição MEF2 , Camundongos , Músculo Esquelético/fisiologia , Proteína MyoD/genética , Fatores de Regulação Miogênica , Miogenina/biossíntese , Cadeias Leves de Miosina/metabolismo , Comunicação Parácrina/fisiologia , Isoformas de Proteínas/biossíntese , Fatores de Transcrição/genética , Transcrição Gênica/fisiologia
8.
Int J Oncol ; 47(5): 1703-10, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26397460

RESUMO

The homeobox transcription factor orthodenticle homeobox 2 (OTX2) plays a critical role in very early neurogenesis, but can become oncogenic when aberrantly expressed later in life. We previously discovered its novel oncogenic role in the malignant childhood brain tumor medulloblastoma and hypothesize an oncogenic role in retinoblastoma. Primary retinoblastoma tumors and cell lines were analyzed by quantitative-PCR, immunoblotting and immunohistochemistry for OTX2. The effect of modulating OTX2 expression on tumorigenesis was tested pharmacologically and by siRNA. A lentiviral shRNA-engineered vector was used for conditional knockdown studies on tumor growth in vivo. A luciferase reporter assay was used to analyze ATRA's effect on OTX2's promoter. In this study on retinoblastoma, OTX2 was frequently amplified and/or overexpressed in primary tumors and cell lines. Knockdown of OTX2 expression by siRNA or pharmacologic inhibition by all-trans retinoic acid (ATRA) repressed OTX2 expression and cell proliferation and significantly decreased tumor growth in vivo. Loss of OTX2 expression also resulted in decreased expression of C-MYC and CRX, genes previously implicated in retinoblastoma tumorigenesis. Loss of OTX2 expression increased the phosphorylation of RB, a potential mechanism of modulating cell proliferation. Aberrant expression of OTX2 may contribute to the development of retinoblastoma. OTX2 may serve as a common transcription factor that interlinks multiple tumor-driving pathways. These results also show that OTX2 can be genetically and pharmacologically targeted, providing an exciting new therapeutic option that may be less toxic and more efficacious than current treatments.


Assuntos
Proteínas de Homeodomínio/genética , Fatores de Transcrição Otx/biossíntese , Proteínas Proto-Oncogênicas c-myc/genética , Retinoblastoma/genética , Retinoblastoma/terapia , Transdução de Sinais/genética , Transativadores/genética , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Proteínas de Homeodomínio/biossíntese , Humanos , Fatores de Transcrição Otx/genética , Fosforilação , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/biossíntese , Retinoblastoma/patologia , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Transativadores/biossíntese , Tretinoína/administração & dosagem
9.
Zhonghua Yi Xue Za Zhi ; 82(14): 990-4, 2002 Jul 25.
Artigo em Zh | MEDLINE | ID: mdl-12181096

RESUMO

OBJECTIVE: To investigate the effect of interleukin-1 receptor antagonist gene therapy on type II collagen induced arthritis in DBA/1 mice. METHODS: Plasmid pcDI-IL-1ra that expresses IL-1ra in eukaryotes was constructed by inserting human IL-1ra cDNA into the eukaryotic expression vector pcDI. Eukaryotes were transfected with the plasmid pcDI-IL-1ra in vivo and in vitro. The expression of 8 IL-1ra was examined by ELISA and immunohistochemistry. Type II collagen was used to induced arthritis in 32 DBA/1 mice. This plasmid was injected into the muscles of DBA/1 mice with arthritis induced by type II collagen by gene gun (20 micro g/mouse) and into the muscle of 8 mice by intramuscular injection (200 micro g/mouse). After the administration, the condition of arthritis was observed. The serum IL-ira was examined 6 and 12 days after administration. The expression of IL-ira in muscles was tested by computerized imaging analysis. RESULTS: PCR and DNA sequencing proved the accuracy of the inserted fragment. ELISA and immunohistochemistry detected high expression of IL-ira in vivo and in vitro. The absorbance ( A ) 490 value of IL-ira in the mouse muscle was 0.52 +/- 0.03 in gene gun group, and 0.48 +/- 0.02 in intramuscular injection group, all higher than that in the control group (0.41 +/- 0.02,P < 0.01 and P < 0.05). The serum IL-ira values in the gene gun group and in tramuscular injection group 6 days and 12 days after therapy were all significantly higher than that in the control group (all P < 0.01; and P < 0.01 and P < 0.05). Since the 6 th day after therapy, the redness and swelling of joints in both therapies groups were alleviated. Pathological examination made 12 days after therapy showed relief at different degrees of the infiltration of inflammatory cells, hyperplasia of synovia, bone infiltration, and cartilage destruction, especially in the gene gun group. CONCLUSION: Gene therapy of IL-ira via non-virus eukaryotic expression vactor, especially by gene gun, is effective in treating arthritis induced by type II collagen.


Assuntos
Artrite/terapia , Colágeno Tipo II/imunologia , Terapia Genética , Sialoglicoproteínas/genética , Animais , Proteína Antagonista do Receptor de Interleucina 1 , Masculino , Camundongos , Camundongos Endogâmicos DBA , Sialoglicoproteínas/análise
10.
Int J Oncol ; 44(4): 1243-51, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24481586

RESUMO

Previous studies identified the frequent loss of adherens junction-associated protein 1 (AJAP1) expression in glioblastoma (GBM) and its correlation with worse survival. AJAP1 may suppress glioma cell migration, which plays an important role in tumor progression in malignant gliomas such as GBM. However, the role of AJAP1 in cell cycle arrest or apoptosis and resistance to chemotherapy remains unclear. Based on microarray screening results, quantitative PCR and luciferase plasmid reporter constructs were used to evaluate the possible regulatory role of AJAP1 on MAGEA2 expression and function. Cell death assays, TUNEL and other markers of apoptosis were utilized to detect cell apoptosis. Restoration of AJAP1 expression in glioma cells was analyzed after temozolomide exposure. AJAP1 suppressed the expression of MAGEA2 and inhibited the transcriptional activity of MAGEA2 in glioma cells. As AJAP1 expression decreased MAGEA2 protein expression apoptosis increased moderately. Consistent with increased cell death, the induced loss of MAGEA2 expression correlated with increased caspase 3/7 activity, BCL2/BAX ratio and TUNEL signal. AJAP1 expression enhanced cell death in the presence of temozolomide. This study suggests AJAP1 may also function as a pro-apoptotic factor and potentiate cell death by temozolomide in glioma cells. This effect may be partially explained by AJAP1-mediated gene regulation of MAGEA2.


Assuntos
Apoptose/genética , Neoplasias Encefálicas/patologia , Moléculas de Adesão Celular/genética , Glioblastoma/patologia , Antígenos Específicos de Melanoma/biossíntese , Proteínas de Neoplasias/biossíntese , Junções Aderentes , Antineoplásicos Alquilantes/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Caspase 3/metabolismo , Caspase 7/metabolismo , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Glioblastoma/tratamento farmacológico , Células HEK293 , Histona Desacetilases/metabolismo , Humanos , Marcação In Situ das Extremidades Cortadas , Temozolomida , Transcrição Gênica , Proteína Supressora de Tumor p53/biossíntese , Proteína X Associada a bcl-2/biossíntese
11.
CNS Neurosci Ther ; 20(5): 429-37, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24483339

RESUMO

AIMS: Down-regulation of AJAP1 in glioblastoma multiforme (GBM) has been reported. However, the expression profiles of AJAP1 in gliomas and the underlying mechanisms of AJAP1 function on invasion are still poorly understood. METHODS: The gene profiles of AJAP1 in glioma patients were studied among four independent cohorts. Confocal imaging was used to analyze the AJAP1 localization. After AJAP1 overexpression in GBM cell lines, cellular polarity, cytoskeleton distribution, and antitumor effect were investigated in vitro and in vivo. RESULTS: AJAP1 expression was significantly decreased in gliomas compared with normal brain in REMBRANDT and CGCA cohorts. Additionally, low AJAP1 expression was associated with worse survival in GBMs in REMBRANDT and TCGA U133A cohorts and was significantly associated with classical and mesenchymal subtypes of GBMs among four cohorts. Confocal imaging indicated AJAP1 localized in cell membranes in low-grade gliomas and AJAP1-overexpressing GBM cells, but difficult to assess in high-grade gliomas due to its absence. AJAP1 overexpression altered the cytoskeleton and cellular polarity in vitro and inhibited the tumor growth in vivo. CONCLUSIONS: AJAP1 is dysregulated at an early stage of gliomagenesis and may suppress glioma cell invasion and proliferation, which suggests that AJAP1 may be a potential diagnostic and prognostic marker for gliomas.


Assuntos
Neoplasias Encefálicas/metabolismo , Encéfalo/metabolismo , Moléculas de Adesão Celular/metabolismo , Citoesqueleto/metabolismo , Glioma/metabolismo , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Estudos de Coortes , Citoesqueleto/ultraestrutura , Progressão da Doença , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Glioma/patologia , Humanos , Estimativa de Kaplan-Meier , Camundongos Nus , Microscopia Confocal , Estadiamento de Neoplasias , Transplante de Neoplasias
12.
Mol Cancer Res ; 10(2): 208-17, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22241217

RESUMO

Glioblastoma is universally fatal because of its propensity for rapid recurrence due to highly migratory tumor cells. Unraveling the genomic complexity that underlies this migratory characteristic could provide therapeutic targets that would greatly complement current surgical therapy. Using multiple high-resolution genomic screening methods, we identified a single locus, adherens junctional associated protein 1 (AJAP1) on chromosome 1p36 that is lost or epigenetically silenced in many glioblastomas. We found AJAP1 expression absent or reduced in 86% and 100% of primary glioblastoma tumors and cell lines, respectively, and the loss of expression correlates with AJAP1 methylation. Restoration of AJAP1 gene expression by transfection or demethylation agents results in decreased tumor cell migration in glioblastoma cell lines. This work shows the significant loss of expression of AJAP1 in glioblastoma and provides evidence of its role in the highly migratory characteristic of these tumors.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Moléculas de Adesão Celular/genética , Movimento Celular/genética , Epigênese Genética , Glioblastoma/genética , Glioblastoma/patologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/metabolismo , Proliferação de Células , Metilação de DNA/efeitos dos fármacos , Decitabina , Regulação Neoplásica da Expressão Gênica , Humanos , RNA Interferente Pequeno
13.
Anticancer Agents Med Chem ; 10(7): 543-55, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20950259

RESUMO

Glioblastoma multiforme (GBM) is one of the most common and most aggressive types of primary brain tumors in humans. Even with aggressive surgical resections using state of the art preoperative and intraoperative neuroim-aging, along with the most recent techniques in radiotherapy and chemotherapy, the prognosis for GBM patients remains dismal. Survival after diagnosis is about 12-14 months. The tumor cells which already have migrated into normal brain tissue beyond the surgical resection margin account for the inability to effectively treat this tumor. Understanding how to control the migration of GBM cells is paramount to future therapies. In this review, we will focus on the emerging targets and agents which are being exploited to inhibit the migration of glioma cells in GBM.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Encéfalo/patologia , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Terapia de Alvo Molecular , Antineoplásicos/farmacologia , Neoplasias Encefálicas/terapia , Movimento Celular , Terapia Combinada , Citocinas/metabolismo , Feminino , Glioblastoma/terapia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Masculino , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/fisiologia , Prognóstico , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais
14.
Pharmgenomics Pers Med ; 3: 111-27, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-23226047

RESUMO

Glioblastoma multiforme (GBM) remains one of the most malignant primary central nervous system tumors. Personalized therapeutic approaches have not become standard of care for GBM, but science is fast approaching this goal. GBM's heterogeneous genomic landscape and resistance to radiotherapy and chemotherapy make this tumor one of the most challenging to treat. Recent advances in genome-wide studies and genetic profiling show that there is unlikely to be a single genetic or cellular event that can be effectively targeted in all patients. Instead, future therapies will likely require personalization for each patient's tumor genotype or proteomic profile. Over the past year, many investigations specifically focused simultaneously on strategies to target oncogenic pathways, angiogenesis, tumor immunology, epigenomic events, glioma stem cells (GSCs), and the highly migratory glioma cell population. Combination therapy targeting multiple pathways is becoming a fast growing area of research, and many studies put special attention on small molecule inhibitors. Because GBM is a highly vascular tumor, therapy that directs monoclonal antibodies or small molecule tyrosine kinase inhibitors toward angiogenic factors is also an area of focus for the development of new therapies. Passive, active, and adoptive immunotherapies have been explored by many studies recently, and epigenetic regulation of gene expression with microRNAs is also becoming an important area of study. GSCs can be useful targets to stop tumor recurrence and proliferation, and recent research has found key molecules that regulate GBM cell migration that can be targeted by therapy. Current standard of care for GBM remains nonspecific; however, pharmacogenomic studies are underway to pave the way for patient-specific therapies that are based on the unique aberrant pathways in individual patients. In conclusion, recent studies in GBM have found many diverse molecular targets possible for therapy. The next obstacle in treating this fatal tumor is ascertaining which molecules in each patient should be targeted and how best to target them, so that we can move our current nonspecific therapies toward the realm of personalized medicine.

15.
Neuro Oncol ; 12(9): 956-66, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20472715

RESUMO

Glioblastoma multiforme (GBM) is one of the deadliest tumors afflicting humans, and the mechanisms of its onset and progression remain largely undefined. Our attempts to elucidate its molecular pathogenesis through DNA copy-number analysis by genome-wide digital karyotyping and single nucleotide polymorphism arrays identified a dramatic focal amplification on chromosome 1q32 in 4 of 57 GBM tumors. Quantitative real-time PCR measurements revealed that HDMX is the most commonly amplified and overexpressed gene in the 1q32 locus. Further genetic screening of 284 low- and high-grade gliomas revealed that HDMX amplifications occur solely in pediatric and adult GBMs and that they are mutually exclusive of TP53 mutations and MDM2 amplifications. Here, we demonstrate that HDMX regulates p53 to promote GBM growth and attenuates tumor response to chemotherapy. In GBM cells, HDMX overexpression inhibits p53-mediated transcriptional activation of p21, releases cells from G0 to G1 phase, and enhances cellular proliferation. HDMX overexpression does not affect the expression of PUMA and BAX proapoptotic genes. While in GBM cells treated with the chemotherapeutic agent 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU), HDMX appears to stabilize p53 and promote phosphorylation of the DNA double-stranded break repair protein H2AX, up-regulate the DNA repair gene VPX, stimulate DNA repair, and confer resistance to BCNU. In summary, HDMX exhibits bona fide oncogenic properties and offers a promising molecular target for GBM therapeutic intervention.


Assuntos
Neoplasias Encefálicas/genética , Regulação da Expressão Gênica/genética , Glioblastoma/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Adulto , Animais , Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Carmustina/farmacologia , Proteínas de Ciclo Celular , Criança , Resistencia a Medicamentos Antineoplásicos/genética , Amplificação de Genes , Expressão Gênica , Estudo de Associação Genômica Ampla , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Humanos , Immunoblotting , Hibridização in Situ Fluorescente , Camundongos , Proteínas Nucleares/genética , Polimorfismo de Nucleotídeo Único , Proteínas Proto-Oncogênicas/genética , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Cancer Res ; 70(1): 181-91, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20028867

RESUMO

OTX2 is a developmentally regulated transcription factor involved in early morphogenesis of the central nervous system. This gene is amplified and overexpressed in medulloblastoma cell lines, but the nature and extent of its genetic alterations in primary tumors have not been evaluated. Analysis of a large cohort of primary medulloblastomas revealed frequent focal copy number gain of a region minimally containing OTX2 as a single gene. OTX2 copy number gain was restricted to tumor subtypes that did not express a molecular signature of Wnt or Shh pathway activation. FISH analysis revealed copy number gain in a subset of cells within medulloblastoma samples, suggesting a late event in tumor progression. Gain of OTX2 copy number was associated with the presence of anaplastic histologic features and shorter survival in medulloblastoma patients. In support of a functional role, ectopic OTX2 expression enhanced proliferation and tumorigenicity of immortalized primary cells, whereas OTX2 knockdown in medulloblastoma cells prolonged the survival of animals bearing xenograft tumors. Mechanistic investigations revealed upregulation of MYC as a potential mechanism whereby OTX2 promotes tumor progression. Our findings define OTX2 as an important oncogenic driver in medulloblastoma.


Assuntos
Neoplasias Cerebelares/genética , Regulação Neoplásica da Expressão Gênica , Meduloblastoma/genética , Fatores de Transcrição Otx/genética , Animais , Western Blotting , Neoplasias Cerebelares/metabolismo , Progressão da Doença , Dosagem de Genes , Genes myc/genética , Proteínas Hedgehog/genética , Humanos , Hibridização in Situ Fluorescente , Meduloblastoma/metabolismo , Camundongos , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Polimorfismo de Nucleotídeo Único
17.
Expert Opin Investig Drugs ; 18(8): 1061-83, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19555299

RESUMO

Malignant gliomas such as glioblastoma multiforme (GBM) present some of the greatest challenges in the management of cancer patients worldwide, despite notable recent achievements in oncology. Even with aggressive surgical resections using state-of-the-art preoperative and intraoperative neuroimaging, along with recent advances in radiotherapy and chemotherapy, the prognosis for GBM patients remains dismal: median survival after diagnosis is about 14 months. Established good prognostic factors are limited, but include young age, high Karnofsky Performance Status (KPS), high mini-mental status examination score, O6-methylguanine methyltransferase promoter methylation, and resection of > 98% of the tumor. Standard treatment includes resection, followed by concurrent chemotherapy and radiotherapy. GBM research is being conducted worldwide at a remarkable pace, with some of the more recent promising studies focused on identification of aberrant genetic events and signaling pathways, tumor stem cell identification and characterization, modulation of tumor immunological responses, combination therapies, and understanding of the rare long-term survivors. Past treatment strategies have failed for various reasons; however, newer strategies in trials today and on the horizon encourage optimism. To help illustrate 'where we have been' with this fatal disease and 'where we are going' with contemporary studies, we include in this review a detailed history of Phase III clinical trials for GBM, with a final emphasis on exciting new treatment strategies that offer hope for future GBM therapy.


Assuntos
Neoplasias do Sistema Nervoso Central/terapia , Glioblastoma/terapia , Antineoplásicos/uso terapêutico , Neoplasias do Sistema Nervoso Central/epidemiologia , Neoplasias do Sistema Nervoso Central/imunologia , Neoplasias do Sistema Nervoso Central/patologia , Ensaios Clínicos Fase III como Assunto , Terapia Combinada , Glioblastoma/epidemiologia , Glioblastoma/imunologia , Glioblastoma/patologia , Humanos , Procedimentos Neurocirúrgicos , Radioterapia , Resultado do Tratamento
18.
Expert Opin Ther Targets ; 13(6): 701-18, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19409033

RESUMO

Glioblastoma is the commonest primary brain tumor, as well as the deadliest. Malignant gliomas such as glioblastoma multiforme (GBM) present some of the greatest challenges in the management of cancer patients worldwide, despite notable recent achievements in oncology. Even with aggressive surgical resections using state-of-the-art preoperative and intraoperative neuroimaging, along with recent advances in radiotherapy and chemotherapy, the prognosis for GBM patients remains dismal: survival after diagnosis is about 1 year. Established prognostic factors are limited, but include age, Karnofsky performance status, mini-mental status examination score, O6-methylguanine methyltransferase promoter methylation and extent of surgery. Standard treatment includes resection of > 95% of the tumor, followed by concurrent chemotherapy and radiotherapy. Nevertheless, GBM research is being conducted worldwide at a remarkable pace, in the laboratory and at the bedside, with some of the more recent promising studies focused on identification of aberrant genetic events and signaling pathways to develop molecular-based targeted therapies, tumor stem cell identification and characterization, modulation of tumor immunological responses and understanding of the rare long-term survivors. With this universally fatal disease, any small breakthrough will have a significant impact on survival and provide hope to the thousands of patients who receive this diagnosis annually. This review describes the epidemiology, clinical presentation, pathology and tumor immunology, with a focus on understanding the molecular biology that underlies the current targeted therapeutics being tested.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/genética , Terapia Combinada , Glioblastoma/diagnóstico , Glioblastoma/genética , Humanos , Prognóstico , Fatores de Risco
19.
Clin Med Oncol ; 3: 39-52, 2009 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-19777070

RESUMO

In the adult population, glioblastoma multiforme is one of the most common primary brain tumors encountered. Unfortunately, this highly malignant tumor represents over 50% of all types of primary central nervous system gliomas. The vast majority of GBMs develops quite rapidly without clinical, radiological, or morphologic evidence of a less malignant precursor lesion (primary or de novo GBMs), as compared to secondary GBMs that develop slowly by progression from diffuse low-grade astrocytomas. These GBM subtypes must be kept in mind because they may constitute distinct disease entities. Even though they look histologically quite similar, they likely involve different genetic alterations and signaling pathways. Decades of surgical therapy, radiotherapy, and chemotherapy have failed to drastically change survival. Clearly, we do not fully understand this tumor; however, the exciting genetic revolution in glioma research over the past decade is providing a promising outlook for exploring this tumor at the genetic level. Science has begun to elucidate the numerous genetic alterations and critical signaling pathways, and it has opened new exciting areas of research such as glioma stem cell biology and neoangiogenesis. This work has already begun to improve our understanding of GBM cell proliferation, migration, and invasion. Indeed, exciting novel targeted therapies are making their way to clinical trials based on this increased knowledge. This review provides the current understanding of GBM oncogenomics, signaling pathways, and glioma stem cell biology and discusses the potential new therapeutic targets on the horizon.

20.
Cancer Res ; 69(23): 9105-11, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19920201

RESUMO

Glioblastoma multiforme is the most prevalent type of adult brain tumor and one of the deadliest tumors known to mankind. The genetic understanding of glioblastoma multiforme is, however, limited, and the molecular mechanisms that facilitate glioblastoma multiforme cell survival and growth within the tumor microenvironment are largely unknown. We applied digital karyotyping and single nucleotide polymorphism arrays to screen for copy-number changes in glioblastoma multiforme samples and found that the most frequently amplified region is at chromosome 7p11.2. The high resolution of digital karyotyping and single nucleotide polymorphism arrays permits the precise delineation of amplicon boundaries and has enabled identification of the minimal region of amplification at chromosome 7p11.2, which contains two genes, EGFR and SEC61gamma. SEC61gamma encodes a subunit of a heterotrimeric protein channel located in the endoplasmic reticulum (ER). In addition to its high frequency of gene amplification in glioblastoma multiforme, SEC61gamma is also remarkably overexpressed in 77% of glioblastoma multiforme but not in lower-grade gliomas. The small interfering RNA-mediated knockdown of SEC61gamma expression in tumor cells led to growth suppression and apoptosis. Furthermore, we showed that pharmacologic ER stress agents induce SEC61gamma expression in glioblastoma multiforme cells. Together, these results indicate that aberrant expression of SEC61gamma serves significant roles in glioblastoma multiforme cell survival likely via a mechanism that is involved in the cytoprotective ER stress-adaptive response to the tumor microenvironment.


Assuntos
Neoplasias Encefálicas/genética , Retículo Endoplasmático/genética , Glioblastoma/genética , Proteínas de Membrana/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Processos de Crescimento Celular/genética , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/patologia , Receptores ErbB/biossíntese , Receptores ErbB/genética , Amplificação de Genes , Genes erbB-1 , Glioblastoma/metabolismo , Glioblastoma/patologia , Células HeLa , Humanos , Proteínas de Membrana/biossíntese , Proto-Oncogene Mas , RNA Interferente Pequeno/genética , Canais de Translocação SEC , Tunicamicina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA